Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(16): 3350-3367.e19, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37421950

RESUMEN

Synucleinopathies are characterized by the accumulation of α-synuclein (α-Syn) aggregates in the brain. Positron emission tomography (PET) imaging of synucleinopathies requires radiopharmaceuticals that selectively bind α-Syn deposits. We report the identification of a brain permeable and rapid washout PET tracer [18F]-F0502B, which shows high binding affinity for α-Syn, but not for Aß or Tau fibrils, and preferential binding to α-Syn aggregates in the brain sections. Employing several cycles of counter screenings with in vitro fibrils, intraneuronal aggregates, and neurodegenerative disease brain sections from several mice models and human subjects, [18F]-F0502B images α-Syn deposits in the brains of mouse and non-human primate PD models. We further determined the atomic structure of the α-Syn fibril-F0502B complex by cryo-EM and revealed parallel diagonal stacking of F0502B on the fibril surface through an intense noncovalent bonding network via inter-ligand interactions. Therefore, [18F]-F0502B is a promising lead compound for imaging aggregated α-Syn in synucleinopathies.


Asunto(s)
Enfermedades Neurodegenerativas , Sinucleinopatías , Animales , Humanos , alfa-Sinucleína/metabolismo , Sinucleinopatías/diagnóstico por imagen , Sinucleinopatías/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Tomografía de Emisión de Positrones , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo
2.
Nature ; 603(7901): 470-476, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35236988

RESUMEN

Alzheimer's disease has a higher incidence in older women, with a spike in cognitive decline that tracks with visceral adiposity, dysregulated energy homeostasis and bone loss during the menopausal transition1,2. Inhibiting the action of follicle-stimulating hormone (FSH) reduces body fat, enhances thermogenesis, increases bone mass and lowers serum cholesterol in mice3-7. Here we show that FSH acts directly on hippocampal and cortical neurons to accelerate amyloid-ß and Tau deposition and impair cognition in mice displaying features of Alzheimer's disease. Blocking FSH action in these mice abrogates the Alzheimer's disease-like phenotype by inhibiting the neuronal C/EBPß-δ-secretase pathway. These data not only suggest a causal role for rising serum FSH levels in the exaggerated Alzheimer's disease pathophysiology during menopause, but also reveal an opportunity for treating Alzheimer's disease, obesity, osteoporosis and dyslipidaemia with a single FSH-blocking agent.


Asunto(s)
Enfermedad de Alzheimer , Hormona Folículo Estimulante , Anciano , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Densidad Ósea , Cognición , Femenino , Hormona Folículo Estimulante/metabolismo , Humanos , Ratones , Termogénesis
3.
EMBO J ; 40(3): e105537, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33351190

RESUMEN

The netrin-1/DCC ligand/receptor pair has key roles in central nervous system (CNS) development, mediating axonal, and neuronal navigation. Although expression of netrin-1 and DCC is maintained in the adult brain, little is known about their role in mature neurons. Notably, netrin-1 is highly expressed in the adult substantia nigra, leading us to investigate a role of the netrin-1/DCC pair in adult nigral neuron fate. Here, we show that silencing netrin-1 in the adult substantia nigra of mice induces DCC cleavage and a significant loss of dopamine neurons, resulting in motor deficits. Because loss of adult dopamine neurons and motor impairments are features of Parkinson's disease (PD), we studied the potential impact of netrin-1 in different animal models of PD. We demonstrate that both overexpression of netrin-1 and brain administration of recombinant netrin-1 are neuroprotective and neurorestorative in mouse and rat models of PD. Of interest, we observed that netrin-1 levels are significantly reduced in PD patient brain samples. These results highlight the key role of netrin-1 in adult dopamine neuron fate, and the therapeutic potential of targeting netrin-1 signaling in PD.


Asunto(s)
Receptor DCC/metabolismo , Netrina-1/genética , Netrina-1/metabolismo , Enfermedad de Parkinson/genética , Sustancia Negra/citología , Animales , Muerte Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Regulación hacia Abajo , Femenino , Silenciador del Gen , Humanos , Masculino , Ratones , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Ratas , Transducción de Señal , Sustancia Negra/metabolismo
4.
EMBO J ; 40(17): e106320, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34260075

RESUMEN

Inflammation plays an important role in the pathogenesis of Alzheimer's disease (AD). Some evidence suggests that misfolded protein aggregates found in AD brains may have originated from the gut, but the mechanism underlying this phenomenon is not fully understood. C/EBPß/δ-secretase signaling in the colon was investigated in a 3xTg AD mouse model in an age-dependent manner. We applied chronic administration of 1% dextran sodium sulfate (DSS) to trigger gut leakage or colonic injection of Aß or Tau fibrils or AD patient brain lysates in 3xTg mice and combined it with excision/cutting of the gut-brain connecting vagus nerve (vagotomy), in order to explore the role of the gut-brain axis in the development of AD-like pathologies and to monitor C/EBPß/δ-secretase signaling under those conditions. We found that C/EBPß/δ-secretase signaling is temporally activated in the gut of AD patients and 3xTg mice, initiating formation of Aß and Tau fibrils that spread to the brain. DSS treatment promotes gut leakage and facilitates AD-like pathologies in both the gut and the brain of 3xTg mice in a C/EBPß/δ-secretase-dependent manner. Vagotomy selectively blunts this signaling, attenuates Aß and Tau pathologies, and restores learning and memory. Aß or Tau fibrils or AD patient brain lysates injected into the colon propagate from the gut into the brain via the vagus nerve, triggering AD pathology and cognitive dysfunction. The results indicate that inflammation activates C/EBPß/δ-secretase and initiates AD-associated pathologies in the gut, which are subsequently transmitted to the brain via the vagus nerve.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Colitis/metabolismo , Colon/metabolismo , Proteínas tau/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Cisteína Endopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
Mol Psychiatry ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658772

RESUMEN

Early onset familial Alzheimer's disease (FAD) with APP, PS1/2 (presenilins) mutation accounts for only a small portion of AD cases, and most are late-onset sporadic. However, majority of AD mouse models are developed to mimic the genetic cause of human AD by overexpressing mutated forms of human APP, PS1/2, and/or Tau protein, though there is no Tau mutation in AD, and no single mouse model recapitulates all aspects of AD pathology. Here, we report Thy1-ApoE4/C/EBPß double transgenic mouse model that demonstrates key AD pathologies in an age-dependent manner in absence of any human APP or PS1/2 mutation. Using the clinical diagnosis criteria, we show that this mouse model exhibits tempo-spatial features in AD patient brains, including progressive cognitive decline associated with brain atrophy, which is accompanied with extensive neuronal degeneration. Remarkably, the mice display gradual Aß aggregation and neurofibrillary tangles formation in the brain validated by Aß PET and Tau PET. Moreover, the mice reveal widespread neuroinflammation as shown in AD brains. Hence, Thy1-ApoE4/C/EBPß mouse model acts as a sporadic AD mouse model, reconstituting the major AD pathologies.

6.
Mol Cell ; 67(5): 812-825.e5, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28826672

RESUMEN

Delta-secretase, a lysosomal asparagine endopeptidase (AEP), simultaneously cleaves both APP and tau, controlling the onset of pathogenesis of Alzheimer's disease (AD). However, how this protease is post-translationally regulated remains unclear. Here we report that serine-arginine protein kinase 2 (SRPK2) phosphorylates delta-secretase and enhances its enzymatic activity. SRPK2 phosphorylates serine 226 on delta-secretase and accelerates its autocatalytic cleavage, leading to its cytoplasmic translocation and escalated enzymatic activities. Delta-secretase is highly phosphorylated in human AD brains, tightly correlated with SRPK2 activity. Overexpression of a phosphorylation mimetic (S226D) in young 3xTg mice strongly promotes APP and tau fragmentation and facilitates amyloid plaque deposits and neurofibrillary tangle (NFT) formation, resulting in cognitive impairment. Conversely, viral injection of the non-phosphorylatable mutant (S226A) into 5XFAD mice decreases APP and tau proteolytic cleavage, attenuates AD pathologies, and reverses cognitive defects. Our findings support that delta-secretase phosphorylation by SRPK2 plays a critical role in aggravating AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Encéfalo/enzimología , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/psicología , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Conducta Animal , Encéfalo/patología , Encéfalo/fisiopatología , Cognición , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Células HEK293 , Células HeLa , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Fenotipo , Fosforilación , Placa Amiloide , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Interferencia de ARN , Serina , Especificidad por Sustrato , Factores de Tiempo , Transfección , Proteínas tau/genética , Proteínas tau/metabolismo
7.
Mol Psychiatry ; 28(3): 1337-1350, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36543925

RESUMEN

Gut dysbiosis contributes to Parkinson's disease (PD) pathogenesis. Gastrointestinal disturbances in PD patients, along with gut leakage and intestinal inflammation, take place long before motor disorders. However, it remains unknown what bacterial species in gut microbiomes play the key role in driving PD pathogenesis. Here we show that Helicobacter hepaticus (H. hepaticus), abundant in gut microbiota from rotenone-treated human α-Synuclein gene (SNCA) transgenic mice and PD patients, initiates α-Synuclein pathology and motor deficits in an AEP-dependent manner in SNCA mice. Chronic Dextran sodium sulfate (DSS) treatment, an inflammatory inducer in the gut, activates AEP (asparagine endopeptidase) that cleaves α-Synuclein N103 and triggers its aggregation, promoting inflammation in the gut and the brain and motor defects in SNCA mice. PD fecal microbiota transplant or live H. hepaticus administration into antibiotics cocktail (Abx)-pretreated SNCA mice induces α-Synuclein pathology, inflammation in the gut and brain, and motor dysfunctions, for which AEP is indispensable. Hence, Helicobacter hepaticus enriched in PD gut microbiomes may facilitate α-Synuclein pathologies and motor impairments via activating AEP.


Asunto(s)
Trastornos Motores , Enfermedad de Parkinson , Ratones , Humanos , Animales , Enfermedad de Parkinson/genética , alfa-Sinucleína , Helicobacter hepaticus , Ratones Transgénicos , Dopamina , Inflamación
8.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34140411

RESUMEN

The molecular mechanism of Alzheimer's disease (AD) pathogenesis remains obscure. Life and/or environmental events, such as traumatic brain injury (TBI), high-fat diet (HFD), and chronic cerebral hypoperfusion (CCH), are proposed exogenous risk factors for AD. BDNF/TrkB, an essential neurotrophic signaling for synaptic plasticity and neuronal survival, are reduced in the aged brain and in AD patients. Here, we show that environmental factors activate C/EBPß, an inflammatory transcription factor, which subsequently up-regulates δ-secretase that simultaneously cleaves both APP and Tau, triggering AD neuropathological changes. These adverse effects are additively exacerbated in BDNF+/- or TrkB+/- mice. Strikingly, TBI provokes both senile plaque deposit and neurofibrillary tangles (NFT) formation in TrkB+/- mice, associated with augmented neuroinflammation and extensive neuronal loss, leading to cognitive deficits. Depletion of C/EBPß inhibits TBI-induced AD-like pathologies in these mice. Remarkably, amyloid aggregates and NFT are tempospatially distributed in TrkB+/- mice brains after TBI, providing insight into their spreading in the progression of AD-like pathologies. Hence, our study revealed the roles of exogenous (TBI, HFD, and CCH) and endogenous (TrkB/BDNF) risk factors in the onset of AD-associated pathologies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Progresión de la Enfermedad , Ambiente , Factores de Crecimiento Nervioso/metabolismo , Transducción de Señal , Envejecimiento/metabolismo , Enfermedad de Alzheimer/complicaciones , Amiloide/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/patología , Cisteína Endopeptidasas/metabolismo , Dieta Alta en Grasa , Humanos , Ratones Endogámicos C57BL , Ovillos Neurofibrilares/patología , Placa Amiloide/patología , Receptor trkB/metabolismo , Factores de Riesgo
9.
Eur J Neurosci ; 58(6): 3555-3568, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37608574

RESUMEN

Limited axon regeneration following peripheral nerve injury may be related to activation of the lysosomal protease, asparaginyl endopeptidase (AEP, δ-secretase) and its degradation of the microtubule associated protein, Tau. Activity of AEP was increased at the site of sciatic nerve transection and repair but blocked in mice treated systemically with a specific AEP inhibitor, compound 11 (CP11). Treatments with CP11 enhanced axon regeneration in vivo. Amplitudes of compound muscle action potentials recorded 4 weeks after nerve transection and repair and 2 weeks after daily treatments with CP11 were double those of vehicle-treated mice. At that time after injury, axons of significantly more motor and sensory neurons had regenerated successfully and reinnervated the tibialis anterior and gastrocnemius muscles in CP11-treated mice than vehicle-treated controls. In cultured adult dorsal root ganglion neurons derived from wild type mice that were treated in vitro for 24 h with CP11, neurites were nearly 50% longer than in vehicle-treated controls and similar to neurite lengths in cultures treated with the TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF). Combined treatment with CP11 and 7,8-DHF did not enhance outgrowth more than treatments with either one alone. Enhanced neurite outgrowth produced by CP11 was found also in the presence of the TrkB inhibitor, ANA-12, indicating that the enhancement was independent of TrkB signalling. Longer neurites were found after CP11 treatment in both TrkB+ and TrkB- neurons. Delta secretase inhibition by CP11 is a treatment for peripheral nerve injury with great potential.


Asunto(s)
Axones , Traumatismos de los Nervios Periféricos , Animales , Ratones , Secretasas de la Proteína Precursora del Amiloide , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Regeneración Nerviosa , Neuritas
10.
Mol Psychiatry ; 27(8): 3396-3409, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35546632

RESUMEN

Diabetes is a risk factor for Alzheimer's disease (AD), which is also called type 3 diabetes with insulin reduction and insulin resistance in AD patient brains. However, the molecular mechanism coupling diabetes to AD onset remains incompletely understood. Here we show that inflammation, associated with obesity and diabetes elicited by high-fat diet (HFD), activates neuronal C/EBPß/AEP signaling that drives AD pathologies and cognitive disorders. HFD stimulates diabetes and insulin resistance in neuronal Thy1-C/EBPß transgenic (Tg) mice, accompanied with prominent mouse Aß accumulation and hyperphosphorylated Tau aggregation in the brain, triggering cognitive deficits. These effects are profoundly diminished when AEP is deleted from C/EBPß Tg mice. Chronic treatment with inflammatory lipopolysaccharide (LPS) facilitates AD pathologies and cognitive disorders in C/EBPß Tg but not in wild-type mice, and these deleterious effects were substantially alleviated in C/EBPß Tg/AEP -/- mice. Remarkably, the anti-inflammatory drug aspirin strongly attenuates HFD-induced diabetes and AD pathologies in neuronal C/EBPß Tg mice. Therefore, our findings demonstrate that inflammation-activated neuronal C/EBPß/AEP signaling couples diabetes to AD.


Asunto(s)
Enfermedad de Alzheimer , Diabetes Mellitus , Resistencia a la Insulina , Animales , Ratones , Enfermedad de Alzheimer/metabolismo , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/fisiología , Ratones Transgénicos , Inflamación/metabolismo , Encéfalo/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diabetes Mellitus/metabolismo , Modelos Animales de Enfermedad
11.
Mol Psychiatry ; 27(7): 3034-3046, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35422468

RESUMEN

Atherosclerosis (ATH) and Alzheimer's disease (AD) are both age-dependent inflammatory diseases, associated with infiltrated macrophages and vascular pathology and overlapping molecules. C/EBPß, an Aß or inflammatory cytokine-activated transcription factor, and AEP (asparagine endopeptidase) are intimately implicated in both ATH and AD; however, whether C/EBPß/AEP signaling couples ATH to AD pathogenesis remains incompletely understood. Here we show that C/EBPß/AEP pathway mediates ATH pathology and couples ATH to AD. Deletion of C/EBPß or AEP from primary macrophages diminishes cholesterol load, and inactivation of this pathway reduces foam cell formation and lesions in aorta in ApoE-/- mice, fed with HFD (high-fat-diet). Knockout of ApoE from 3xTg AD mouse model augments serum LDL and increases lesion areas in the aorta. Depletion of C/EBPß or AEP from 3xTg/ApoE-/- mice substantially attenuates these effects and elevates cerebral blood flow and vessel length, improving cognitive functions. Strikingly, knockdown of ApoE from the hippocampus of 3xTg mice decreases the cerebral blood flow and vessel length and aggravates AD pathologies, leading to cognitive deficits. Inactivation of C/EBPß/AEP pathway alleviates these events and restores cognitive functions. Hence, our findings demonstrate that C/EBPß/AEP signaling couples ATH to AD via mediating vascular pathology.


Asunto(s)
Enfermedad de Alzheimer , Aterosclerosis , Proteína beta Potenciadora de Unión a CCAAT , Enfermedad de Alzheimer/metabolismo , Animales , Aterosclerosis/complicaciones , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados para ApoE
12.
Proc Natl Acad Sci U S A ; 117(39): 24503-24513, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32929029

RESUMEN

The Hippo (MST1/2) pathway plays a critical role in restricting tissue growth in adults and modulating cell proliferation, differentiation, and migration in developing organs. Netrin1, a secreted laminin-related protein, is essential for nervous system development. However, the mechanisms underlying MST1 regulation by the extrinsic signals remain unclear. Here, we demonstrate that Netrin1 reduction in Parkinson's disease (PD) activates MST1, which selectively binds and phosphorylates netrin receptor UNC5B on T428 residue, promoting its apoptotic activation and dopaminergic neuronal loss. Netrin1 deprivation stimulates MST1 activation and interaction with UNC5B, diminishing YAP levels and escalating cell deaths. Knockout of UNC5B abolishes netrin depletion-induced dopaminergic loss, whereas blockade of MST1 phosphorylating UNC5B suppresses neuronal apoptosis. Remarkably, Netrin1 is reduced in PD patient brains, associated with MST1 activation and UNC5B T428 phosphorylation, which is accompanied by YAP reduction and apoptotic activation. Hence, Netrin1 regulates Hippo (MST1) pathway in dopaminergic neuronal loss in PD via UNC5B receptor.


Asunto(s)
Apoptosis , Neuronas Dopaminérgicas/citología , Receptores de Netrina/metabolismo , Netrina-1/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Proliferación Celular , Neuronas Dopaminérgicas/metabolismo , Humanos , Ratones , Ratones Noqueados , Receptores de Netrina/química , Receptores de Netrina/genética , Netrina-1/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/fisiopatología , Fosforilación , Proteínas Serina-Treonina Quinasas/genética
13.
Gut ; 71(11): 2233-2252, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017199

RESUMEN

OBJECTIVE: This study is to investigate the role of gut dysbiosis in triggering inflammation in the brain and its contribution to Alzheimer's disease (AD) pathogenesis. DESIGN: We analysed the gut microbiota composition of 3×Tg mice in an age-dependent manner. We generated germ-free 3×Tg mice and recolonisation of germ-free 3×Tg mice with fecal samples from both patients with AD and age-matched healthy donors. RESULTS: Microbial 16S rRNA sequencing revealed Bacteroides enrichment. We found a prominent reduction of cerebral amyloid-ß plaques and neurofibrillary tangles pathology in germ-free 3×Tg mice as compared with specific-pathogen-free mice. And hippocampal RNAseq showed that inflammatory pathway and insulin/IGF-1 signalling in 3×Tg mice brain are aberrantly altered in the absence of gut microbiota. Poly-unsaturated fatty acid metabolites identified by metabolomic analysis, and their oxidative enzymes were selectively elevated, corresponding with microglia activation and inflammation. AD patients' gut microbiome exacerbated AD pathologies in 3×Tg mice, associated with C/EBPß/asparagine endopeptidase pathway activation and cognitive dysfunctions compared with healthy donors' microbiota transplants. CONCLUSIONS: These findings support that a complex gut microbiome is required for behavioural defects, microglia activation and AD pathologies, the gut microbiome contributes to pathologies in an AD mouse model and that dysbiosis of the human microbiome might be a risk factor for AD.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Insulinas , Enfermedad de Alzheimer/metabolismo , Animales , Cognición , Modelos Animales de Enfermedad , Disbiosis , Ácidos Grasos Insaturados , Microbioma Gastrointestinal/fisiología , Humanos , Inflamación/metabolismo , Factor I del Crecimiento Similar a la Insulina , Ratones , Enfermedades Neuroinflamatorias , Placa Amiloide/patología , ARN Ribosómico 16S
14.
EMBO J ; 37(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29769405

RESUMEN

Dopaminergic neurodegeneration in Parkinson's disease (PD) is associated with abnormal dopamine metabolism by MAO-B (monoamine oxidase-B) and intracellular α-Synuclein (α-Syn) aggregates, called the Lewy body. However, the molecular relationship between α-Syn and MAO-B remains unclear. Here, we show that α-Syn directly binds to MAO-B and stimulates its enzymatic activity, which triggers AEP (asparagine endopeptidase; legumain) activation and subsequent α-Syn cleavage at N103, leading to dopaminergic neurodegeneration. Interestingly, the dopamine metabolite, DOPAL, strongly activates AEP, and the N103 fragment of α-Syn binds and activates MAO-B. Accordingly, overexpression of AEP in SNCA transgenic mice elicits α-Syn N103 cleavage and accelerates PD pathogenesis, and inhibition of MAO-B by Rasagiline diminishes α-Syn-mediated PD pathology and motor dysfunction. Moreover, virally mediated expression of α-Syn N103 induces PD pathogenesis in wild-type, but not MAO-B-null mice. Our findings thus support that AEP-mediated cleavage of α-Syn at N103 is required for the association and activation of MAO-B, mediating PD pathogenesis.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Monoaminooxidasa/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animales , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Dopamina/genética , Dopamina/metabolismo , Indanos/farmacología , Ratones , Ratones Transgénicos , Monoaminooxidasa/genética , Inhibidores de la Monoaminooxidasa/farmacología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , alfa-Sinucleína/genética
15.
Mov Disord ; 37(9): 1817-1830, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36054165

RESUMEN

BACKGROUND: The deposition of α-synuclein (α-Syn) in the brain is the pathological hallmark of Parkinson's disease (PD). Epidemiological data indicate that exposure to fine particulate matter (≤2.5 µm in aerodynamic diameter [PM2.5]) is associated with an increased risk for PD. OBJECTIVE: The aim of this study is to investigate whether PM2.5 has a direct effect on α-Syn pathology and how it drives the risk for PD. METHODS: PM2.5 was added into α-Syn monomers and different cell models to test whether PM2.5 can promote the fibrillization and aggregation of α-Syn. α-Syn A53T transgenic mice and α-Syn knockout mice were used to investigate the effects of PM2.5 on PD-like pathology. RESULTS: PM2.5 triggers the fibrillization of α-Syn and promotes the formation of α-Syn fibrils with enhanced seeding activity and neurotoxicity. PM2.5 also induces mitochondrial dysfunction and oxidative stress. Intrastriatal injection or intranasal administration of PM2.5 exacerbates α-Syn pathology and dopaminergic neuronal degeneration in α-Syn A53T transgenic mice. The detrimental effect of PM2.5 was attenuated in α-Syn knockout mice. CONCLUSIONS: Our results identify that PM2.5 exposure could promote the α-Syn pathology, providing mechanistic insights into how PM2.5 increases the risk for PD. © 2022 International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , Ratones , Ratones Noqueados , Ratones Transgénicos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Material Particulado/toxicidad , alfa-Sinucleína/genética
16.
Mol Psychiatry ; 26(7): 2943-2963, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32782380

RESUMEN

Neurotrophins promote neuronal survival and synaptic plasticity via activating the tropomyosin receptor kinases. BDNF and its high-affinity receptor TrkB are reduced in Alzheimer's disease (AD), contributing to progressive cognitive decline. However, how the signaling mediates AD pathologies remains incompletely understood. Here we show that the TrkB receptor binds and phosphorylates APP, reducing amyloid-ß production, which are abrogated by δ-secretase cleavage of TrkB in AD. Remarkably, BDNF stimulates TrkB to phosphorylate APP Y687 residue that accumulates APP in the TGN (Trans-Golgi Network) and diminishes its amyloidogenic cleavage. Delta-secretase cleaves TrkB at N365 and N486/489 residues and abolishes its neurotrophic activity, decreasing p-APP Y687 and altering its subcellular trafficking. Notably, both TrkB and APP are robustly cleaved by δ-secretase in AD brains, accompanied by mitigated TrkB signaling and reduced p-Y687. Blockade of TrkB cleavage attenuates AD pathologies in 5xFAD mice, rescuing the learning and memory. Viral expression of TrkB 1-486 fragment in the hippocampus of APP/PS1 mice facilitates amyloid pathology and mitigates cognitive functions. Hence, δ-secretase cleaves TrkB and blunts its phosphorylation of APP, facilitating AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Glicoproteínas de Membrana , Ratones , Fosforilación , Proteínas Tirosina Quinasas , Receptor trkB/metabolismo
17.
Mol Psychiatry ; 26(10): 6002-6022, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33339957

RESUMEN

The apolipoprotein E ε4 (APOE4) allele is a major genetic risk factor for Alzheimer's disease (AD), and its protein product, ApoE4, exerts its deleterious effects mainly by influencing amyloid-ß (Aß) and Tau (neurofibrillary tangles, NFTs) deposition in the brain. However, the molecular mechanism dictating its expression during ageing and in AD remains incompletely clear. Here we show that C/EBPß acts as a pivotal transcription factor for APOE and mediates its mRNA levels in an age-dependent manner. C/EBPß binds the promoter of APOE and escalates its expression in the brain. Knockout of C/EBPß in AD mouse models diminishes ApoE expression and Aß pathologies, whereas overexpression of C/EBPß accelerates AD pathologies, which can be attenuated by anti-ApoE monoclonal antibody or deletion of ApoE via its specific shRNA. Remarkably, C/EBPß selectively promotes more ApoE4 expression versus ApoE3 in human neurons, correlating with higher activation of C/EBPß in human AD brains with ApoE4/4 compared to ApoE3/3. Therefore, our data support that C/EBPß is a crucial transcription factor for temporally regulating APOE gene expression, modulating ApoE4's role in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E4/genética , Apolipoproteínas E , Encéfalo/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Ratones , Ratones Noqueados
18.
Mol Psychiatry ; 26(2): 568-585, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32086435

RESUMEN

Parkinson's disease (PD) is characterized by dopaminergic neuronal loss and the presence of intra-neuronal Lewy body (LB) inclusions with aggregated α-synuclein (α-Syn) as the major component. MAOB, a crucial monoamine oxidase for dopamine metabolism, triggers oxidative stress in dopaminergic neurons and α-Syn aggregation. However, the key molecular mechanism that mediates PD pathogenesis remains elusive. Here we show that C/EBPß acts as an age-dependent transcription factor for both α-Syn and MAOB, and initiates the PD pathologies by upregulating these two pivotal players, in addition to escalating δ-secretase activity to cleave α-Syn and promotes its neurotoxicity. Overexpression of C/EBPß in human wild-type α-Syn transgenic mice facilitates PD pathologies and elicits motor disorders associated with augmentation of δ-secretase, α-Syn, and MAOB. In contrast, depletion of C/EBPß from human α-Syn Tg mice abolishes rotenone-elicited PD pathologies and motor impairments via downregulating the expression of these key factors. Hence, our study supports that C/EBPß/δ-secretase signaling mediates PD pathogenesis via regulating the expression and cleavage of α-Syn and MAOB.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Secretasas de la Proteína Precursora del Amiloide , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Neuronas Dopaminérgicas , Ratones , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética
19.
Mol Psychiatry ; 26(12): 7838-7850, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34489530

RESUMEN

Respiratory chain complex I deficiency elicits mitochondrial dysfunction and reactive oxidative species (ROS), which plays a crucial role in Parkinson's disease (PD) pathogenesis. However, it remains unclear whether the impairment in other complexes in the mitochondrial oxidative phosphorylation chain is also sufficient to trigger PD onset. Here we show that inhibition of Complex II or III in the electron transport chain (ETC) induces the motor disorder and PD pathologies in neuronal Thy1-C/EBPß transgenic mice. Through a cell-based screening of mitochondrial respiratory chain inhibitors, we identified TTFA (complex II inhibitor) and Atovaquone (complex III inhibitor), which robustly block the oxidative phosphorylation functions, strongly escalate ROS, and activate C/EBPß/AEP pathway that triggers dopaminergic neuronal cell death. Oral administration of these inhibitors to Thy1-C/EBPß mice elicits constipation and motor defects, associated with Lewy body-like inclusions. Deletion of SDHD (Succinate dehydrogenase) gene from the complex II in the Substantia Nigra of Thy1-C/EBPß mice triggers ROS and PD pathologies, resulting in motor disorders. Hence, our findings demonstrate that mitochondrial ETC inactivation triggers PD pathogenesis via activating C/EBPß/AEP pathway.


Asunto(s)
Enfermedad de Parkinson , Animales , Neuronas Dopaminérgicas/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Sustancia Negra/patología
20.
Mol Psychiatry ; 26(2): 586-603, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-30382187

RESUMEN

δ-Secretase, an age-dependent asparagine protease, cleaves both amyloid precursor protein (APP) and Tau and is required for amyloid plaque and neurofibrillary tangle pathologies in Alzheimer's disease (AD). However, whether δ-secretase activation is sufficient to trigger AD pathogenesis remains unknown. Here we show that the fragments of δ-secretase-cleavage, APP (586-695) and Tau(1-368), additively drive AD pathogenesis and cognitive dysfunctions. Tau(1-368) strongly augments BACE1 expression and Aß generation in the presence of APP. The Tau(1-368) fragment is more robust than full-length Tau in binding active STAT1, a BACE1 transcription factor, and promotes its nuclear translocation, upregulating BACE1 and Aß production. Notably, Aß-activated SGK1 or JAK2 kinase phosphorylates STAT1 and induces its association with Tau(1-368). Inhibition of these kinases diminishes stimulatory effect of Tau(1-368). Knockout of STAT1 abolishes AD pathologies induced by δ-secretase-generated APP and Tau fragments. Thus, we show that Tau may not only be a downstream effector of Aß in the amyloid hypothesis, but also act as a driving force for Aß, when cleaved by δ-secretase.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Humanos , Ovillos Neurofibrilares , Factor de Transcripción STAT1 , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA